1
|
James J, Fokin AI, Guschin DY, Wang H, Polesskaya A, Rubtsova SN, Clainche CL, Silberzan P, Gautreau AM, Romero S. Vinculin-Arp2/3 interaction inhibits branched actin assembly to control migration and proliferation. Life Sci Alliance 2025; 8:e202402583. [PMID: 39547716 PMCID: PMC11568829 DOI: 10.26508/lsa.202402583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Vinculin is a mechanotransducer that reinforces links between cell adhesions and linear arrays of actin filaments upon myosin-mediated contractility. Both adhesions to the substratum and neighboring cells, however, are initiated within membrane protrusions that originate from Arp2/3-nucleated branched actin networks. Vinculin has been reported to interact with the Arp2/3 complex, but the role of this interaction remains poorly understood. Here, we compared the phenotypes of vinculin knock-out (KO) cells with those of knock-in (KI-P878A) cells, where the point mutation P878A that impairs the Arp2/3 interaction is introduced in the two vinculin alleles of MCF10A mammary epithelial cells. The interaction of vinculin with Arp2/3 inhibits actin polymerization at membrane protrusions and decreases migration persistence of single cells. In cell monolayers, vinculin recruits Arp2/3 and the vinculin-Arp2/3 interaction participates in cell-cell junction plasticity. Through this interaction, vinculin controls the decision to enter a new cell cycle as a function of cell density.
Collapse
Affiliation(s)
- John James
- Laboratory of Structural Biology of the Cell (BIOC), CNRS UMR7654, École Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Artem I Fokin
- Laboratory of Structural Biology of the Cell (BIOC), CNRS UMR7654, École Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
- Laboratoire PhysicoChimie Curie UMR168, Institut Curie, Paris Sciences et Lettres, Centre National de la Recherche Scientifique, Sorbonne Université, Paris, France
| | - Dmitry Y Guschin
- Laboratory of Structural Biology of the Cell (BIOC), CNRS UMR7654, École Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Hong Wang
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Anna Polesskaya
- Laboratory of Structural Biology of the Cell (BIOC), CNRS UMR7654, École Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Svetlana N Rubtsova
- Laboratory of Structural Biology of the Cell (BIOC), CNRS UMR7654, École Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Christophe Le Clainche
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Pascal Silberzan
- Laboratoire PhysicoChimie Curie UMR168, Institut Curie, Paris Sciences et Lettres, Centre National de la Recherche Scientifique, Sorbonne Université, Paris, France
| | - Alexis M Gautreau
- Laboratory of Structural Biology of the Cell (BIOC), CNRS UMR7654, École Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Stéphane Romero
- Laboratory of Structural Biology of the Cell (BIOC), CNRS UMR7654, École Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
2
|
Blob A, Ventzke D, Rölleke U, Nies G, Munk A, Schaedel L, Köster S. Global alignment and local curvature of microtubules in mouse fibroblasts are robust against perturbations of vimentin and actin. SOFT MATTER 2025; 21:641-651. [PMID: 39749806 PMCID: PMC11697242 DOI: 10.1039/d4sm01127a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
The eukaryotic cytoskeleton is an intricate network of three types of mechanically distinct biopolymers - actin filaments, microtubules and intermediate filaments (IFs). These filamentous networks determine essential cellular functions and properties. Among them, microtubules are important for intracellular transport and establishing cell polarity during migration. Despite their intrinsic stiffness, they exhibit characteristic bending and buckling in cells due to nonthermal forces acting on them. Interactions between cytoskeletal filaments have been found but are complex and diverse with respect to their effect on the mechanical behavior of the filaments and the architecture of networks. We systematically study how actin and vimentin IFs influence the network structure and local bending of microtubules by analyzing fluorescence microscopy images of mouse fibroblasts on protein micropatterns. Our automated analysis averages over large amounts of data to mitigate the effect of the considerable natural variance in biological cell data. We find that the radial orientation of microtubules in circular cells is robust and is established independently of vimentin and actin networks. Observing the local curvature of microtubules, we find highly similar average bending of microtubules in the entire cell regardless of the cytoskeletal surrounding. Small systematic differences cannot be attributed directly to vimentin and actin densities. Our results suggest that, on average, microtubules in unpolarized mouse fibroblasts are unexpectedly independent of the rest of the cytoskeleton in their global network structure and their local curvature.
Collapse
Affiliation(s)
- Anna Blob
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany.
| | - David Ventzke
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany.
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077 Göttingen, Germany
| | - Ulrike Rölleke
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Germany
| | - Giacomo Nies
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077 Göttingen, Germany
| | - Axel Munk
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077 Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany
| | - Laura Schaedel
- Department of Physics, Center for Biophysics, Saarland University, Campus A2 4, 66123 Saarbrücken, Germany
| | - Sarah Köster
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany
| |
Collapse
|
3
|
Yoshihara S, Nakata T, Kashiwazaki J, Aoyama K, Mabuchi I. In Vitro Formation of Actin Ring in the Fission Yeast Cell Extracts. Cytoskeleton (Hoboken) 2025. [PMID: 39835694 DOI: 10.1002/cm.21997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/18/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
Cytokinesis in animal and fungal cells requires the contraction of actomyosin-based contractile rings formed in the division cortex of the cell during late mitosis. However, the detailed mechanism remains incompletely understood. Here, we aim to develop a novel cell-free system by encapsulating cell extracts obtained from fission yeast cells within lipid vesicles, which subsequently leads to the formation of a contractile ring-like structure inside the vesicles. Using this system, we found that an actin ring structure formed in vesicles of a size similar to that of fission yeast cells, with the frequency of ring appearance increasing in the presence of PI(4,5)P2 (PIP2). In contrast, larger vesicles tended to form actin bundles, which were sometimes associated with ring structures or network-like structures. The effects of various inhibitors affecting cytoskeleton formation were investigated, revealing that actin polymerization was essential for the formation of these actin structures. Additionally, the involvement of ATP, the Schizosaccharomyces pombe PLK "Plo1," and the small GTPase Rho was suggested to play a crucial role in this process. Examination of mitotic extracts revealed the formation of actin dot structures in phosphatidylethanolamine vesicles. However, most of these structures disappeared in the presence of PIP2, leading to the formation of actin Rings instead. Using extracts from cells expressing α-actinin Ain1 or myosin-II light chain Rlc1, both fused with fluorescent proteins, we found that these proteins colocalized with actin bundles. In summary, we have developed a new semi-in vitro system to investigate mechanisms such as cell division and cytoskeleton formation.
Collapse
Affiliation(s)
- Shogo Yoshihara
- Department of Cell Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- The Center for Brain Integration Research (CBIR), TMDU, Tokyo, Japan
- Department of Life Science, Faculty of Science, Gakushuin University, Mejiro, Tokyo, Japan
| | - Takao Nakata
- Department of Cell Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- The Center for Brain Integration Research (CBIR), TMDU, Tokyo, Japan
| | - Jun Kashiwazaki
- Department of Life Science, Faculty of Science, Gakushuin University, Mejiro, Tokyo, Japan
- Radioisotope Division, Research Facility Center for Science and Technology, Kobe University, Kobe, Hyogo, Japan
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Hyogo, Japan
| | - Kazuhiro Aoyama
- NanoPort Japan, Application Laboratory, Thermo Fisher Scientific, Tokyo, Japan
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, Osaka, Japan
| | - Issei Mabuchi
- Department of Life Science, Faculty of Science, Gakushuin University, Mejiro, Tokyo, Japan
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
- Institute of Human Culture Studies, Otsuma Women's University, Tokyo, Japan
| |
Collapse
|
4
|
Gong R, Reynolds MJ, Carney KR, Hamilton K, Bidone TC, Alushin GM. Fascin structural plasticity mediates flexible actin bundle construction. Nat Struct Mol Biol 2025:10.1038/s41594-024-01477-2. [PMID: 39833469 DOI: 10.1038/s41594-024-01477-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025]
Abstract
Fascin cross-links actin filaments (F-actin) into bundles that support tubular membrane protrusions including filopodia and stereocilia. Fascin dysregulation drives aberrant cell migration during metastasis, and fascin inhibitors are under development as cancer therapeutics. Here, we use cryo-EM, cryo-electron tomography coupled with custom denoising and computational modeling to probe human fascin-1's F-actin cross-linking mechanisms across spatial scales. Our fascin cross-bridge structure reveals an asymmetric F-actin binding conformation that is allosterically blocked by the inhibitor G2. Reconstructions of seven-filament hexagonal bundle elements, variability analysis and simulations show how structural plasticity enables fascin to bridge varied interfilament orientations, accommodating mismatches between F-actin's helical symmetry and bundle hexagonal packing. Tomography of many-filament bundles and modeling uncover geometric rules underlying emergent fascin binding patterns, as well as the accumulation of unfavorable cross-links that limit bundle size. Collectively, this work shows how fascin harnesses fine-tuned nanoscale structural dynamics to build and regulate micron-scale F-actin bundles.
Collapse
Affiliation(s)
- Rui Gong
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA.
| | - Matthew J Reynolds
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Keith R Carney
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Keith Hamilton
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Tamara C Bidone
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Gregory M Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
5
|
Jani P, Colville MJ, Park S, Ha Y, Paszek MJ, Abbott NL. Influence of the glycocalyx on the size and mechanical properties of plasma membrane-derived vesicles. SOFT MATTER 2025; 21:463-475. [PMID: 39717887 PMCID: PMC11667464 DOI: 10.1039/d4sm01317d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 11/30/2024] [Indexed: 12/25/2024]
Abstract
Recent studies have reported that the overexpression of MUC1 glycoproteins on cell surfaces changes the morphology of cell plasma membranes and increases the blebbing of vesicles from them, supporting the hypothesis that entropic forces exerted by MUC1 change the spontaneous curvature of cell membranes. However, how MUC1 is incorporated into and influences the size and biophysical properties of plasma-membrane-blebbed vesicles is not understood. Here we report single-vesicle-level characterization of giant plasma membrane vesicles (GPMVs) derived from cells overexpressing MUC1, revealing a 40× variation in MUC1 density between GPMVs from a single preparation and a strong correlation between GPMV size and MUC1 density. By dispersing GPMVs in aqueous liquid crystals (LCs), we show that the elasticity of the LC can be used to strain individual GPMVs into spindle-like shapes, consistent with the straining of fluid-like membranes. To quantify the influence of MUC1 on membrane mechanical properties, we analyze the shapes of strained GPMVs within a theoretical framework that integrates the effects of MUC1 density and GPMV size on strain. We measure the spontaneous curvature of GPMV membranes to be 2-10 μm-1 and weakly influenced by the 40× variation in MUC1 density, a conclusion we validate by performing independent experiments in which MUC1 is enzymatically removed from GPMVs. Overall, our study advances the understanding of heterogeneity in size and MUC1 density in GPMVs, and establishes single-vesicle-level methods for characterization of mechanical properties within a heterogeneous population of GPMVs. Furthermore, our measurements highlight differences between membrane properties of GPMVs and their parent cells.
Collapse
Affiliation(s)
- Purvil Jani
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA.
| | - Marshall J Colville
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA.
- Field of Biophysics, Cornell University, Ithaca, NY 14853, USA
| | - Sangwoo Park
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA.
- Field of Biophysics, Cornell University, Ithaca, NY 14853, USA
| | - Youlim Ha
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA.
| | - Matthew J Paszek
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA.
- Field of Biophysics, Cornell University, Ithaca, NY 14853, USA
- Nancy E. and Peter C. Meining School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Nicholas L Abbott
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
6
|
Wang Y, Wang Y, Zhu Y, Yu P, Zhou F, Zhang A, Gu Y, Jin R, Li J, Zheng F, Yu A, Ye D, Xu Y, Liu YJ, Saw TB, Hu G, Lim CT, Yu FX. Angiomotin cleavage promotes leader formation and collective cell migration. Dev Cell 2025; 60:101-118.e7. [PMID: 39389053 DOI: 10.1016/j.devcel.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 05/22/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024]
Abstract
Collective cell migration (CCM) is involved in multiple biological processes, including embryonic morphogenesis, angiogenesis, and cancer invasion. However, the molecular mechanisms underlying CCM, especially leader cell formation, are poorly understood. Here, we show that a signaling pathway regulating angiomotin (AMOT) cleavage plays a role in CCM, using mammalian epithelial cells and mouse models. In a confluent epithelial monolayer, full-length AMOT localizes at cell-cell junctions and limits cell motility. After cleavage, the C-terminal fragment of AMOT (AMOT-CT) translocates to the cell-matrix interface to promote the maturation of focal adhesions (FAs), generate traction force, and induce leader cell formation. Meanwhile, decreased full-length AMOT at cell-cell junctions leads to tissue fluidization and coherent migration of cell collectives. Hence, the cleavage of AMOT serves as a molecular switch to generate polarized contraction, promoting leader cell formation and CCM.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yebin Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yuwen Zhu
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Pengcheng Yu
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Fanhui Zhou
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Anlan Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yuan Gu
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Ruxin Jin
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Jin Li
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Fengyun Zheng
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Aijuan Yu
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Dan Ye
- Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yanhui Xu
- Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yan-Jun Liu
- Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Thuan Beng Saw
- Research Center for Industries of the Future and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China; Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore; Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Guohong Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore; Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore 117599, Singapore
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, The State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China.
| |
Collapse
|
7
|
Guérin C, N'Diaye AB, Gressin L, Mogilner A, Théry M, Blanchoin L, Colin A. Balancing limited resources in actin network competition. Curr Biol 2025:S0960-9822(24)01634-8. [PMID: 39793569 DOI: 10.1016/j.cub.2024.11.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/05/2024] [Accepted: 11/26/2024] [Indexed: 01/13/2025]
Abstract
In cells, multiple actin networks coexist in a dynamic manner. These networks compete for a common pool of actin monomers and actin-binding proteins. Interestingly, all of these networks manage to coexist despite the strong competition for resources. Moreover, the coexistence of networks with various strengths is key to cell adaptation to external changes. However, a comprehensive view of how these networks coexist in this competitive environment, where resources are limited, is still lacking. To address this question, we used a reconstituted system, in closed microwells, consisting of beads propelled by actin polymerization or micropatterns functionalized with lipids capable of initiating polymerization close to a membrane. This system enabled us to build dynamic actin architectures, competing for a limited pool of proteins, over a period of hours. We demonstrated the importance of protein turnover for the coexistence of actin networks, showing that it ensures resource distribution between weak and strong networks. However, when competition becomes too intense, turnover alone is insufficient, leading to a selection process that favors the strongest networks. Consequently, we emphasize the importance of competition strength, which is defined by the turnover rate, the amount of available protein, and the number of competing structures. More generally, this work illustrates how turnover allows biological populations with various competition strengths to coexist despite resource constraints.
Collapse
Affiliation(s)
- Christophe Guérin
- Cytomorpholab, Laboratoire de Physiologie Cellulaire and Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, 17 avenue des Martyrs, 38054 Grenoble, France
| | - Anne-Betty N'Diaye
- Cytomorpholab, Laboratoire de Physiologie Cellulaire and Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, 17 avenue des Martyrs, 38054 Grenoble, France
| | - Laurène Gressin
- Cytomorpholab, Laboratoire de Physiologie Cellulaire and Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, 17 avenue des Martyrs, 38054 Grenoble, France
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences and Department of Biology, New York University, 251 Mercer Street, New York, NY 10012, USA
| | - Manuel Théry
- Cytomorpholab, Institut Chimie Biologie Innovation, Institut Pierre-Gilles de Gennes, Université Paris Sciences et Lettres, CEA, ESPCI, 6 rue Jean Calvin, 75005 Paris, France.
| | - Laurent Blanchoin
- Cytomorpholab, Laboratoire de Physiologie Cellulaire and Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, 17 avenue des Martyrs, 38054 Grenoble, France; Cytomorpholab, Institut Chimie Biologie Innovation, Institut Pierre-Gilles de Gennes, Université Paris Sciences et Lettres, CEA, ESPCI, 6 rue Jean Calvin, 75005 Paris, France.
| | - Alexandra Colin
- Cytomorpholab, Laboratoire de Physiologie Cellulaire and Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, 17 avenue des Martyrs, 38054 Grenoble, France.
| |
Collapse
|
8
|
Casanova-Sepúlveda G, Boggon TJ. Regulation and signaling of the LIM domain kinases. Bioessays 2025; 47:e2400184. [PMID: 39361252 DOI: 10.1002/bies.202400184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 11/17/2024]
Abstract
The LIM domain kinases (LIMKs) are important actin cytoskeleton regulators. These proteins, LIMK1 and LIMK2, are nodes downstream of Rho GTPases and are the key enzymes that phosphorylate cofilin/actin depolymerization factors to regulate filament severing. They therefore perform an essential role in cascades that control actin depolymerization. Signaling of the LIMKs is carefully regulated by numerous inter- and intra-molecular mechanisms. In this review, we discuss recent findings that improve the understanding of LIM domain kinase regulation mechanisms. We also provide an up-to-date review of the role of the LIM domain kinases, their architectural features, how activity is impacted by other proteins, and the implications of these findings for human health and disease.
Collapse
Affiliation(s)
| | - Titus J Boggon
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
- Department of Pharmacology, Yale University, New Haven, Connecticut, USA
- Yale Cancer Center, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
9
|
Rassier DE, Månsson A. Mechanisms of myosin II force generation: insights from novel experimental techniques and approaches. Physiol Rev 2025; 105:1-93. [PMID: 38451233 DOI: 10.1152/physrev.00014.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024] Open
Abstract
Myosin II is a molecular motor that converts chemical energy derived from ATP hydrolysis into mechanical work. Myosin II isoforms are responsible for muscle contraction and a range of cell functions relying on the development of force and motion. When the motor attaches to actin, ATP is hydrolyzed and inorganic phosphate (Pi) and ADP are released from its active site. These reactions are coordinated with changes in the structure of myosin, promoting the so-called "power stroke" that causes the sliding of actin filaments. The general features of the myosin-actin interactions are well accepted, but there are critical issues that remain poorly understood, mostly due to technological limitations. In recent years, there has been a significant advance in structural, biochemical, and mechanical methods that have advanced the field considerably. New modeling approaches have also allowed researchers to understand actomyosin interactions at different levels of analysis. This paper reviews recent studies looking into the interaction between myosin II and actin filaments, which leads to power stroke and force generation. It reviews studies conducted with single myosin molecules, myosins working in filaments, muscle sarcomeres, myofibrils, and fibers. It also reviews the mathematical models that have been used to understand the mechanics of myosin II in approaches focusing on single molecules to ensembles. Finally, it includes brief sections on translational aspects, how changes in the myosin motor by mutations and/or posttranslational modifications may cause detrimental effects in diseases and aging, among other conditions, and how myosin II has become an emerging drug target.
Collapse
Affiliation(s)
- Dilson E Rassier
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Alf Månsson
- Physiology, Linnaeus University, Kalmar, Sweden
| |
Collapse
|
10
|
Sinha B, Biswas A, Kaushik S, Soni GV. Cellular and Nuclear Forces: An Overview. Methods Mol Biol 2025; 2881:3-39. [PMID: 39704936 DOI: 10.1007/978-1-0716-4280-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Biological cells sample their surrounding microenvironments using nanoscale force sensors on the cell surfaces. These surface-based force and stress sensors generate physical and chemical responses inside the cell. The inherently well-connected cytoskeleton and its physical contacts with the force elements on the nuclear membrane lead these physicochemical responses to cascade all the way inside the cell nucleus, physically altering the nuclear state. These physical alterations of the cell nucleus, through yet-unknown complex steps, elicit physical and functional responses from the chromatin in the form of altered gene expression profiles. This mechanism of force/stress sensing by the cell and then its nuclear response has been shown to play a vital role in maintaining robust cellular homeostasis, controlling gene expression profiles during developmental phases as well as cell differentiation. In the last few years, there has been appreciable progress toward the identification of the molecular players responsible for force sensing. However, the actual sensing mechanism of cell surface-bound force sensors and more importantly cascading of the signals, both physical (via cytosolic force sensing elements such as microtubule and actin framework) as well as chemical (cascade of biochemical signaling from cell surface to nuclear surface and further to the chromatin), inside the cell is poorly understood. In this chapter, we present a review of the currently known molecular players in cellular as well as nuclear force sensing repertoire and their possible mechanistic aspects. We also introduce various biophysical concepts and review some frequently used techniques that are used to describe the force/stress sensing and response of a cell. We hope that this will help in asking clearer questions and designing pointed experiments for better understanding of the force-dependent design principles of the cell surface, nuclear surface, and gene expression.
Collapse
Affiliation(s)
- Bidisha Sinha
- Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Arikta Biswas
- Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | | | - Gautam V Soni
- Raman Research Institute, Bangalore, Karnataka, India.
| |
Collapse
|
11
|
Kroll KL, Sosnick TR, Rock RS. Design and Use of AsLOV2-Based Optogenetic Tools for Actin Imaging. Methods Mol Biol 2025; 2840:89-100. [PMID: 39724346 DOI: 10.1007/978-1-0716-4047-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
We present protocols for using an optogenetic tool called LILAC for actin imaging. LILAC is a light-controlled version of Lifeact that uses the Avena sativa LOV2 (AsLOV2) domain. By significantly reducing Lifeact's affinity for the cytoskeleton in the dark, LILAC reduces concentration-dependent negative side effects while enabling new image processing methods. We discuss the considerations for using this probe of live-cell actin dynamics, including fluorescent protein selection, cell maintenance, microscopy protocols, and image processing. Our work highlights the potential of AsLOV2-based optogenetics for novel imaging and control tools in cell biology.
Collapse
Affiliation(s)
- Kourtney L Kroll
- Department of Biochemistry and Molecular Biology & The Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
| | - Tobin R Sosnick
- Department of Biochemistry and Molecular Biology & The Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
| | - Ronald S Rock
- Department of Biochemistry and Molecular Biology & The Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
12
|
George A, Alva E, Brancaleon L, Marucho M. Dynamic and electrophoretic light scattering measurements on microtubules at low concentrations. PLoS One 2024; 19:e0312430. [PMID: 39739655 PMCID: PMC11687780 DOI: 10.1371/journal.pone.0312430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/07/2024] [Indexed: 01/02/2025] Open
Abstract
The accurate characterization of microtubules is essential for understanding their roles in various biological activities in eukaryotic cellular processes. In vitro, experimental data on these systems often need more details and information on sample preparation protocols and experimental techniques. This deficiency leads to unreproducible or contradictory outcomes. The use of diverse experimental methods and preparations yields different results of hydrodynamic and electro-mechanical properties, complicating the process of obtaining meaningful data and conclusive information. This article presents a robust and detailed protocol for performing dynamic light scattering (DLS) and electrophoretic light scattering (ELS) measurements on microtubules at low concentrations. This method ensures accurate and reproducible results on essential microtubule filament parameters such as the diffusion coefficient (D) and electrophoretic mobility (μ) from which other structures' hydrodynamics, electrical, and stability properties can be elucidated.
Collapse
Affiliation(s)
- Annitta George
- Department of Physics and Astronomy, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Ernesto Alva
- Department of Physics and Astronomy, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Lorenzo Brancaleon
- Department of Physics and Astronomy, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Marcelo Marucho
- Department of Physics and Astronomy, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
13
|
Zhang WJ, Pi XW, Le YG, Li TZ. Role of P2X7 receptor in the progression and clinicopathological characteristics of gastric cancer. Sci Rep 2024; 14:31673. [PMID: 39738256 DOI: 10.1038/s41598-024-81515-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
P2X7 receptor (P2X7R) plays a role in regulating tumor progression, but it is unclear whether P2X7R affects the pathological characteristics of patients with gastric cancer and the activity of gastric cancer cells. Therefore, this study preliminarily investigated the relationship between P2X7R and clinicopathological features of patients with gastric cancer, and further explored the effect of P2X7R on the proliferation, migration and invasion of gastric cancer cells through functional experiments. The results showed that P2X7R was highly expressed in gastric cancer tissues and gastric cancer cells. High expression of P2X7R was closely related to lymphatic metastasis, vascular invasion and Tumor-Node-Metastasis (TNM) stage in patients with gastric cancer. High expression of P2X7R predicted poor overall survival in patients. Moreover, the activation of P2X7R by ATP and its analogue BzATP increased the calcium current of gastric cancer cells, enhanced YF actin stress and cell viability, and promoted the proliferation, migration and invasion of gastric cancer cells. While P2X7R antagonists (A438079 and AZD9056) inhibited the proliferation, migration and invasion of gastric cancer cells induced by ATP. Therefore, the data obtained in this experiment suggest that P2X7R may be another potential molecular target for the prevention and treatment of gastric cancer.
Collapse
Affiliation(s)
- Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang city, Jiangxi province, China
| | - Xiong-Wei Pi
- Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang city, Jiangxi province, China
| | - Yi-Guan Le
- Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang city, Jiangxi province, China
| | - Teng-Zheng Li
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang city, Jiangxi province, China.
| |
Collapse
|
14
|
Mishra A, Taylor H, Patil AJ, Mann S. Dynamic Co-Clustering and Self-Sorting in Interactive Protocell Populations. Angew Chem Int Ed Engl 2024:e202420209. [PMID: 39714324 DOI: 10.1002/anie.202420209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/23/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
The design and implementation of collective actions in model protocell communities is an on-going challenge in synthetic protobiology. Herein, we covalently graft alginate or chitosan onto the outer surface of semipermeable enzyme-containing silica colloidosomes to produce hairy catalytic protocells with pH-switchable membrane surface charge. Binary populations of the enzymatically active protocells exhibit self-initiated stimulus-responsive changes in spatial organization such that the mixed community undergoes alternative modes of electrostatically induced self-sorting and reversible co-clustering. We demonstrate that co-clustering, but not self-sorting, mitigates signal attenuation in a binary community of enzyme-containing sender and receiver protocells due to increased proximity effects. The level of signal attenuation is correlated with a time-dependent pH-mediated switch in the spatial organization of the sender and receiver populations. Our results pave the way towards the development of programmable networks of adaptive life-like objects and could have implications for the development of interactive cytomimetic materials and agent-based robotics.
Collapse
Affiliation(s)
- Ananya Mishra
- Centre for Protolife Research and Centre for Organized Matter Chemistry, School of Chemistry, University of Bristol, Bristol, BS8 1TS, United Kingdom
- Max Planck-Bristol Centre for Minimal Biology, School of Chemistry, University of Bristol, Bristol, BS8 1TS, United Kingdom
| | - Hannah Taylor
- Centre for Protolife Research and Centre for Organized Matter Chemistry, School of Chemistry, University of Bristol, Bristol, BS8 1TS, United Kingdom
| | - Avinash J Patil
- Centre for Protolife Research and Centre for Organized Matter Chemistry, School of Chemistry, University of Bristol, Bristol, BS8 1TS, United Kingdom
| | - Stephen Mann
- Centre for Protolife Research and Centre for Organized Matter Chemistry, School of Chemistry, University of Bristol, Bristol, BS8 1TS, United Kingdom
- Max Planck-Bristol Centre for Minimal Biology, School of Chemistry, University of Bristol, Bristol, BS8 1TS, United Kingdom
| |
Collapse
|
15
|
Yang W, Wang Y, Liu G, Wang Y, Wu C. TPM4 condensates glycolytic enzymes and facilitates actin reorganization under hyperosmotic stress. Cell Discov 2024; 10:120. [PMID: 39622827 PMCID: PMC11612400 DOI: 10.1038/s41421-024-00744-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/20/2024] [Indexed: 12/06/2024] Open
Abstract
Actin homeostasis is fundamental for cell structure and consumes a large portion of cellular ATP. It has been documented in the literature that certain glycolytic enzymes can interact with actin, indicating an intricate interplay between the cytoskeleton and cellular metabolism. Here we report that hyperosmotic stress triggers actin severing and subsequent phase separation of the actin-binding protein tropomyosin 4 (TPM4). TPM4 condensates recruit glycolytic enzymes such as HK2, PFKM, and PKM2, while wetting actin filaments. Notably, the condensates of TPM4 and glycolytic enzymes are enriched of NADH and ATP, suggestive of their functional importance in cell metabolism. At cellular level, actin filament assembly is enhanced upon hyperosmotic stress and TPM4 condensation, while depletion of TPM4 impairs osmolarity-induced actin reorganization. At tissue level, colocalized condensates of TPM4 and glycolytic enzymes are observed in renal tissues subjected to hyperosmotic stress. Together, our findings suggest that stress-induced actin perturbation may act on TPM4 to organize glycolytic hubs that tether energy production to cytoskeletal reorganization.
Collapse
Affiliation(s)
- Wenzhong Yang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Yuan Wang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Geyao Liu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Yan Wang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- International Cancer Institute, Peking University, Beijing, China
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- International Cancer Institute, Peking University, Beijing, China.
- Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Beijing, China.
| |
Collapse
|
16
|
Peng Y, Chen B. Role of cell membrane homeostasis in the pathogenicity of pathogenic filamentous fungi. Virulence 2024; 15:2299183. [PMID: 38156783 PMCID: PMC10761126 DOI: 10.1080/21505594.2023.2299183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024] Open
Abstract
The cell membrane forms a fundamental part of all living cells and participates in a variety of physiological processes, such as material exchange, stress response, cell recognition, signal transduction, cellular immunity, apoptosis, and pathogenicity. Here, we review the mechanisms and functions of the membrane structure (lipid components of the membrane and the biosynthesis of unsaturated fatty acids), membrane proteins (transmembrane proteins and proteins contributing to membrane curvature), transcriptional regulation, and cell wall components that influence the virulence and pathogenicity of filamentous fungi.
Collapse
Affiliation(s)
- Yuejin Peng
- Yunnan State Key Laboratory of Conservation and Utilization of Biological Resources, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Bin Chen
- Yunnan State Key Laboratory of Conservation and Utilization of Biological Resources, Yunnan Agricultural University, Kunming, Yunnan, China
| |
Collapse
|
17
|
Sun ZG, Murrell M. Cofilin-Mediated Filament Softening and Crosslinking Counterbalance to Enhance Actin Network Flexibility. PHYSICAL REVIEW LETTERS 2024; 133:218402. [PMID: 39642486 DOI: 10.1103/physrevlett.133.218402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/30/2024] [Indexed: 12/09/2024]
Abstract
Filamentous-actin (F-actin) crosslinking within the cell cytoskeleton mediates the transmission of mechanical forces, enabling changes in cell shape, as occurs during cell division and cell migration. Crosslinking by actin binding proteins (ABPs) generally increases the connectivity of the F-actin network, but also increases network rigidity. As a result, there is a narrow range in the concentration of crosslinker protein at which F-actin networks are both connected and labile. Another ABP, cofilin, severs F-actin filaments at high pH through increasing their bending flexibility and concentrating mechanical stress, inducing fragmentation. By contrast, at lower pH, cofilin increases filament flexibility yet does not sever. Instead, it forms disulfide bonds, which crosslink F-actin into bundles, and bundles into networks. Here, we combine light microscopy and rheology to determine the impact of two potentially opposing effects on the mechanics of F-actin networks-increased flexibility at the filament level, and increased connectivity at the network level. Indeed, by linear rheology, we find that these mechanisms are counterbalanced, such that cofilactin network moduli are only weakly dependent on cofilin concentration over a broad range, in contrast to the dramatic stiffening that occurs with F-actin crosslinking protein. Further, by nonlinear rheology, the network stiffens at a higher stress than crosslinking protein, indicative of a broader range in which the material remains flexible. These results may enable F-actin networks to increase connectivity without heavy penalties to rigidity, and thus provide a new route to modulating active polymer mechanics unseen using traditional F-actin accessory proteins.
Collapse
Affiliation(s)
- Zachary Gao Sun
- Department of Physics, Yale University, 217 Prospect Street, New Haven, Connecticut 06511, USA
- Systems Biology Institute, Yale University, West Haven, Connecticut 06516, USA
- Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, Connecticut 06520, USA
| | - Michael Murrell
- Department of Physics, Yale University, 217 Prospect Street, New Haven, Connecticut 06511, USA
- Systems Biology Institute, Yale University, West Haven, Connecticut 06516, USA
- Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, Connecticut 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
18
|
Pinchiaroli J, Saldanha R, Patteson AE, Robertson-Anderson RM, Gurmessa BJ. Scale-dependent interactions enable emergent microrheological stress response of actin-vimentin composites. SOFT MATTER 2024; 20:9007-9021. [PMID: 39495192 DOI: 10.1039/d4sm00988f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
The mechanical properties of the mammalian cell regulate many cellular functions and are largely dictated by the cytoskeleton, a composite network of protein filaments, including actin, microtubules, and intermediate filaments. Interactions between these distinct filaments give rise to emergent mechanical properties that are difficult to generate synthetically, and recent studies have made great strides in advancing our understanding of the mechanical interplay between actin and microtubule filaments. While intermediate filaments play critical roles in the stress response of cells, their effect on the rheological properties of the composite cytoskeleton remains poorly understood. Here, we use optical tweezers microrheology to measure the linear viscoelastic properties and nonlinear stress response of composites of actin and vimentin with varying molar ratios of actin to vimentin. We reveal a surprising, nearly opposite effect of actin-vimentin network mechanics compared to single-component networks in the linear versus nonlinear regimes. Namely, the linear elastic plateau modulus and zero-shear viscosity are markedly reduced in composites compared to single-component networks of actin or vimentin, whereas the initial response force and stiffness are maximized in composites versus single-component networks in the nonlinear regime. While these emergent trends are indicative of distinct interactions between actin and vimentin, nonlinear stiffening and long-time stress response appear to both be dictated primarily by actin, at odds with previous bulk rheology studies. We demonstrate that these complex, scale-dependent effects arise from the varied contributions of network density, filament stiffness, non-specific interactions, and poroelasticity to the mechanical response at different spatiotemporal scales. Cells may harness this complex behavior to facilitate distinct stress responses at different scales and in response to different stimuli to allow for their hallmark multifunctionality.
Collapse
Affiliation(s)
- Julie Pinchiaroli
- Department of Physics and Astronomy, Bucknell University, Lewisburg, PA 17837, USA.
| | - Renita Saldanha
- Department of Physics and BioInspired Institute, Syracuse University, Syracuse, NY 13210, USA
| | - Alison E Patteson
- Department of Physics and BioInspired Institute, Syracuse University, Syracuse, NY 13210, USA
| | | | - Bekele J Gurmessa
- Department of Physics and Astronomy, Bucknell University, Lewisburg, PA 17837, USA.
| |
Collapse
|
19
|
Mooren OL, McConnell P, DeBrecht JD, Jaysingh A, Cooper JA. Reconstitution of Arp2/3-nucleated actin assembly with proteins CP, V-1, and CARMIL. Curr Biol 2024; 34:5173-5186.e4. [PMID: 39437783 PMCID: PMC11576230 DOI: 10.1016/j.cub.2024.09.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Actin polymerization is often associated with membrane proteins containing capping-protein-interacting (CPI) motifs, such as capping protein, Arp2/3, myosin I linker (CARMIL), CD2AP, and WASHCAP/Fam21. CPI motifs bind directly to actin-capping protein (CP), and this interaction weakens the binding of CP to barbed ends of actin filaments, lessening the ability of CP to functionally cap those ends. The protein V-1/myotrophin binds to the F-actin-binding site on CP and sterically blocks CP from binding barbed ends. CPI-motif proteins also weaken the binding between V-1 and CP, which decreases the inhibitory effects of V-1, thereby freeing CP to cap barbed ends. Here, we address the question of whether CPI-motif proteins on a surface analogous to a membrane lead to net activation or inhibition of actin assembly nucleated by Arp2/3 complex. Using reconstitution with purified components, we discovered that CARMIL at the surface promotes and enhances actin assembly, countering the inhibitory effects of V-1 and thus activating CP. The reconstitution involves the presence of an Arp2/3 activator on the surface, along with Arp2/3 complex, V-1, CP, profilin, and actin monomers in solution, recreating key features of cell physiology.
Collapse
Affiliation(s)
- Olivia L Mooren
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Patrick McConnell
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - James D DeBrecht
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Anshuman Jaysingh
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - John A Cooper
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA.
| |
Collapse
|
20
|
Li X, Wang S, Zhang D, Feng Y, Liu Y, Yu W, Cui L, Harkany T, Verkhratsky A, Xia M, Li B. The periaxonal space as a conduit for cerebrospinal fluid flow to peripheral organs. Proc Natl Acad Sci U S A 2024; 121:e2400024121. [PMID: 39485799 PMCID: PMC11551422 DOI: 10.1073/pnas.2400024121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 09/20/2024] [Indexed: 11/03/2024] Open
Abstract
Mechanisms controlling the movement of the cerebrospinal fluid (CSF) toward peripheral nerves are poorly characterized. We found that, in addition to the foramina Magendie and Luschka for CSF flow toward the subarachnoid space and glymphatic system, CSF outflow could also occur along periaxonal spaces (termed "PAS pathway") from the spinal cord to peripheral organs, such as the liver and pancreas. When interrogating the latter route, we found that serotonin, acting through 5-HT2B receptors expressed in ependymocytes that line the central canal, triggered Ca2+ signals to induce polymerization of F-actin, a cytoskeletal protein, to reduce the volume of ependymal cells. This paralleled an increased rate of PAS-mediated CSF redistribution toward peripheral organs. In the liver, CSF was received by hepatic stellate cells. CSF efflux toward peripheral organs through the PAS pathway represents a mechanism dynamically connecting the nervous system with the periphery. Our findings are compatible with the traditional theory of CSF efflux into the glymphatic system to clear metabolic waste from the cerebral parenchyma. Thus, we extend the knowledge of CSF flow and expand the understanding of connectivity between the CNS and peripheral organs.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China Medical University, Shenyang110122, China
- China Medical University Centre of Forensic Investigation, Shenyang110122, China
| | - Siman Wang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China Medical University, Shenyang110122, China
- China Medical University Centre of Forensic Investigation, Shenyang110122, China
| | - Dianjun Zhang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China Medical University, Shenyang110122, China
- China Medical University Centre of Forensic Investigation, Shenyang110122, China
| | - Yuliang Feng
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China Medical University, Shenyang110122, China
- China Medical University Centre of Forensic Investigation, Shenyang110122, China
| | - Yingyu Liu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China Medical University, Shenyang110122, China
- China Medical University Centre of Forensic Investigation, Shenyang110122, China
| | - Weiyang Yu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China Medical University, Shenyang110122, China
- China Medical University Centre of Forensic Investigation, Shenyang110122, China
| | - Lulu Cui
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China Medical University, Shenyang110122, China
- China Medical University Centre of Forensic Investigation, Shenyang110122, China
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna1090, Austria
- Department of Neuroscience, Biomedicum, Karolinska Institutet, Solna17165, Sweden
| | - Alexei Verkhratsky
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang110122, China
- Faculty of Biology, Medicine and Health, The University of Manchester, ManchesterM13 9PL, United Kingdom
- Department of Neurosciences, University of the Basque Country, Leioa48940, Bizkaia, Spain
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, VilniusLT-01102, Lithuania
| | - Maosheng Xia
- Department of Orthopaedics, The First Hospital, China Medical University, Shenyang110002, China
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, China Medical University, Shenyang110122, China
- China Medical University Centre of Forensic Investigation, Shenyang110122, China
| |
Collapse
|
21
|
Ayushman M, Mikos G, Tong X, Sinha S, Lopez-Fuentes E, Jones S, Cai PC, Lee HP, Morrison AJ, Spakowitz A, Heilshorn SC, Sweet-Cordero A, Yang F. Cell tumbling enhances stem cell differentiation in hydrogels via nuclear mechanotransduction. NATURE MATERIALS 2024:10.1038/s41563-024-02038-0. [PMID: 39487316 DOI: 10.1038/s41563-024-02038-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 09/30/2024] [Indexed: 11/04/2024]
Abstract
Cells can deform their local niche in three dimensions via whole-cell movements such as spreading, migration or volume expansion. These behaviours, occurring over hours to days, influence long-term cell fates including differentiation. Here we report a whole-cell movement that occurs in sliding hydrogels at the minutes timescale, termed cell tumbling, characterized by three-dimensional cell dynamics and hydrogel deformation elicited by heightened seconds-to-minutes-scale cytoskeletal and nuclear activity. Studies inhibiting or promoting the cell tumbling of mesenchymal stem cells show that this behaviour enhances differentiation into chondrocytes. Further, it is associated with a decrease in global chromatin accessibility, which is required for enhanced differentiation. Cell tumbling also occurs during differentiation into other lineages and its differentiation-enhancing effects are validated in various hydrogel platforms. Our results establish that cell tumbling is an additional regulator of stem cell differentiation, mediated by rapid niche deformation and nuclear mechanotransduction.
Collapse
Affiliation(s)
- Manish Ayushman
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Georgios Mikos
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Xinming Tong
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sauradeep Sinha
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Eunice Lopez-Fuentes
- Division of Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Sarah Jones
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Pamela C Cai
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Hung-Pang Lee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Andrew Spakowitz
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Alejandro Sweet-Cordero
- Division of Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Fan Yang
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
22
|
Scholz J, Stephan T, Pérez AG, Csiszár A, Hersch N, Fischer LS, Brühmann S, Körber S, Litschko C, Mijanovic L, Kaufmann T, Lange F, Springer R, Pich A, Jakobs S, Peckham M, Tarantola M, Grashoff C, Merkel R, Faix J. Decisive role of mDia-family formins in cell cortex function of highly adherent cells. SCIENCE ADVANCES 2024; 10:eadp5929. [PMID: 39475610 PMCID: PMC11524191 DOI: 10.1126/sciadv.adp5929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/20/2024] [Indexed: 11/02/2024]
Abstract
Cortical formins, pivotal for the assembly of linear actin filaments beneath the membrane, exert only minor effects on unconfined cell migration of weakly and moderately adherent cells. However, their impact on migration and mechanostability of highly adherent cells remains poorly understood. Here, we demonstrate that loss of cortical actin filaments generated by the formins mDia1 and mDia3 drastically compromises cell migration and mechanics in highly adherent fibroblasts. Biophysical analysis of the mechanical properties of the mutant cells revealed a markedly softened cell cortex in the poorly adherent state. Unexpectedly, in the highly adherent state, associated with a hyperstretched morphology with exaggerated focal adhesions and prominent high-strain stress fibers, they exhibited even higher cortical tension compared to control. Notably, misguidance of intracellular forces, frequently accompanied by stress-fiber rupture, culminated in the formation of tension- and contractility-induced macroapertures, which was instantly followed by excessive lamellipodial protrusion at the periphery, providing critical insights into mechanotransduction of mechanically stressed and highly adherent cells.
Collapse
Affiliation(s)
- Jonas Scholz
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Till Stephan
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Aina Gallemí Pérez
- Institute for Dynamics of Complex Systems, Göttingen, Germany
- Max Planck Institute for Dynamics and Self-Organization, Department LFPB, Göttingen, Germany
| | - Agnes Csiszár
- Institute of Biological Information Processing 2: Mechanobiology, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Nils Hersch
- Institute of Biological Information Processing 2: Mechanobiology, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Lisa S. Fischer
- Institute of Integrative Cell Biology and Physiology, University of Münster, Münster, Germany
| | - Stefan Brühmann
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Sarah Körber
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
- HiLIFE Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Christof Litschko
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Lucija Mijanovic
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Thomas Kaufmann
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Felix Lange
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Ronald Springer
- Institute of Biological Information Processing 2: Mechanobiology, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Andreas Pich
- Research Core Unit Proteomics and Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Stefan Jakobs
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Michelle Peckham
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Marco Tarantola
- Institute for Dynamics of Complex Systems, Göttingen, Germany
- Max Planck Institute for Dynamics and Self-Organization, Department LFPB, Göttingen, Germany
| | - Carsten Grashoff
- Institute of Integrative Cell Biology and Physiology, University of Münster, Münster, Germany
| | - Rudolf Merkel
- Institute of Biological Information Processing 2: Mechanobiology, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
23
|
Zhao X, Sun S, Shi Z, He F, Qi G, Li X, Niu Y. Characterization of Cytoskeletal Profilin Genes in Plasticity Elongation of Mesocotyl and Coleoptile of Maize Under Diverse Abiotic Stresses. Int J Mol Sci 2024; 25:11693. [PMID: 39519245 PMCID: PMC11546416 DOI: 10.3390/ijms252111693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The plasticity elongation of mesocotyl (MES) and coleoptile (COL) largely determines the morphology of maize seedlings under abiotic stresses. The profilin (PRF) proteins play a pivotal role in cytoskeleton dynamics and plant development via regulating actin polymerization. However, little is known about whether and how the expression of the ZmPRF gene family regulates MES and COL elongation in maize under adverse abiotic stresses. Here, a total of eight ZmPRF gene members were identified in the maize genome. They were mainly located in the cytoplasm, chloroplast, and mitochondrion, and clearly divided into four classes, based on phylogenetic analysis. Segmental duplication was the main driver for the expansion of ZmPRF genes. Ka/Ks analysis indicated that most ZmPRF genes were intensely purified and selected. Promoter cis-element analysis suggested their potential roles in response to growth and development, stress adaption, hormone response, and light response. The protein-protein interaction network and two independent RNA-sequencing analyses revealed that eight ZmPRF genes and their thirty-seven interacting genes showed varied expression patterns in MES and COL of three maize genotypes under different sowing depths, 24-epibrassinolide application, and light spectral-quality treatments, of which ZmPRF3.3 was a potential core conserved gene for breeding application. Moreover, the quantitative real-time PCR (qRT-PCR) verified that the relative expression levels of most ZmPRF genes in MES and COL under above treatments were significantly correlated with the plasticity elongation of MES and COL in maize. Therefore, these results perform a comprehensive overview of the ZmPRF family and will provide valuable information for the validation of the function of ZmPRF genes in maize development under diverse abiotic stress.
Collapse
Affiliation(s)
- Xiaoqiang Zhao
- State Key Laboratory of Aridland Crop Science, College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China; (S.S.); (Z.S.); (F.H.); (G.Q.); (X.L.); (Y.N.)
| | | | | | | | | | | | | |
Collapse
|
24
|
Hu Q, Zhao Y, Sun WY, Ou Z, Duan W, Qiu Z, Ge Y, Tang D, Chen T, Cheng X, He RR, Wu S, Ju Z. CK-666 protects against ferroptosis and renal ischemia-reperfusion injury through a microfilament-independent mechanism. J Biol Chem 2024; 300:107942. [PMID: 39481596 PMCID: PMC11625328 DOI: 10.1016/j.jbc.2024.107942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
Ferroptosis is a type of regulated cell death driven by iron-dependent accumulation of lipid peroxidation, exhibiting unique morphological changes. While actin microfilaments are crucial for various cellular processes, including morphogenesis, motility, endocytosis, and cell death, their role in ferroptosis remains unclear. Here, our study reveals that actin microfilaments undergo remodeling and disassembly during ferroptosis. Interestingly, inhibitors that target actin microfilament remodeling do not affect cell sensitivity to ferroptosis, with the exception of CK-666 and its structural analog CK-636. Mechanistically, CK-666 attenuates ferroptosis independently of its canonical function in inhibiting the Arp2/3 complex. Further investigation revealed that CK-666 modulates the ferroptotic transcriptome, prevents lipid degradation, and diminishes lipid peroxidation. In addition, CK-666 does not impact the labile iron pool within cells nor does the inhibition of FSP1 impacts its antiferroptosis activity. Notably, the results of DPPH assay and liposome leakage assay suggest that CK-666 mitigates ferroptosis by directly eliminating lipid peroxidation. Importantly, CK-666 significantly ameliorated renal ischemia-reperfusion injury and ferroptosis in renal tissue, underscoring its potential therapeutic impact.
Collapse
Affiliation(s)
- Qian Hu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China.
| | - Yanan Zhao
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Wan-Yang Sun
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility, Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zexian Ou
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Wentao Duan
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Zeyu Qiu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, China
| | - Yuanlong Ge
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Tianfeng Chen
- College of Chemistry and Materials Science, Jinan University, Guangzhou, China
| | - Xiang Cheng
- Department of Hematology, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Rong-Rong He
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility, Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Shu Wu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
25
|
Chen X, Xu S, Chu B, Guo J, Zhang H, Sun S, Song L, Feng XQ. Applying Spatiotemporal Modeling of Cell Dynamics to Accelerate Drug Development. ACS NANO 2024; 18:29311-29336. [PMID: 39420743 DOI: 10.1021/acsnano.4c12599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Cells act as physical computational programs that utilize input signals to orchestrate molecule-level protein-protein interactions (PPIs), generating and responding to forces, ultimately shaping all of the physiological and pathophysiological behaviors. Genome editing and molecule drugs targeting PPIs hold great promise for the treatments of diseases. Linking genes and molecular drugs with protein-performed cellular behaviors is a key yet challenging issue due to the wide range of spatial and temporal scales involved. Building predictive spatiotemporal modeling systems that can describe the dynamic behaviors of cells intervened by genome editing and molecular drugs at the intersection of biology, chemistry, physics, and computer science will greatly accelerate pharmaceutical advances. Here, we review the mechanical roles of cytoskeletal proteins in orchestrating cellular behaviors alongside significant advancements in biophysical modeling while also addressing the limitations in these models. Then, by integrating generative artificial intelligence (AI) with spatiotemporal multiscale biophysical modeling, we propose a computational pipeline for developing virtual cells, which can simulate and evaluate the therapeutic effects of drugs and genome editing technologies on various cell dynamic behaviors and could have broad biomedical applications. Such virtual cell modeling systems might revolutionize modern biomedical engineering by moving most of the painstaking wet-laboratory effort to computer simulations, substantially saving time and alleviating the financial burden for pharmaceutical industries.
Collapse
Affiliation(s)
- Xindong Chen
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
- BioMap, Beijing 100144, China
| | - Shihao Xu
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Bizhu Chu
- School of Pharmacy, Shenzhen University, Shenzhen 518055, China
- Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jing Guo
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Huikai Zhang
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Shuyi Sun
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Le Song
- BioMap, Beijing 100144, China
| | - Xi-Qiao Feng
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| |
Collapse
|
26
|
Soto JS, Neupane C, Kaur M, Pandey V, Wohlschlegel JA, Khakh BS. Astrocyte Gi-GPCR signaling corrects compulsive-like grooming and anxiety-related behaviors in Sapap3 knockout mice. Neuron 2024; 112:3412-3423.e6. [PMID: 39163865 PMCID: PMC11512628 DOI: 10.1016/j.neuron.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/06/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024]
Abstract
Astrocytes are morphologically complex cells that serve essential roles. They are widely implicated in central nervous system (CNS) disorders, with changes in astrocyte morphology and gene expression accompanying disease. In the Sapap3 knockout (KO) mouse model of compulsive and anxiety-related behaviors related to obsessive-compulsive disorder (OCD), striatal astrocytes display reduced morphology and altered actin cytoskeleton and Gi-G-protein-coupled receptor (Gi-GPCR) signaling proteins. Here, we show that normalizing striatal astrocyte morphology, actin cytoskeleton, and essential homeostatic support functions by targeting the astrocyte Gi-GPCR pathway using chemogenetics corrected phenotypes in Sapap3 KO mice, including anxiety-related and compulsive behaviors. Our data portend an astrocytic pharmacological strategy for rescuing phenotypes in brain disorders that include compromised astrocyte morphology and tissue support.
Collapse
Affiliation(s)
- Joselyn S Soto
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA.
| | - Chiranjivi Neupane
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Muskan Kaur
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Vijaya Pandey
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - James A Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA.
| |
Collapse
|
27
|
Lu C, Zeng T, Wang M, Yoshitomi T, Kawazoe N, Yang Y, Chen G. Influence of viscosity on adipogenic and osteogenic differentiation of mesenchymal stem cells during 2D culture. Biomater Sci 2024; 12:5598-5609. [PMID: 39327896 DOI: 10.1039/d4bm00710g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Accumulatively, cellular behaviours triggered by biochemical cues have been widely explored and the focus of research is gradually shifting to biophysical cues. Compared to physical parameters such as stiffness, substrate morphology and viscoelasticity, the influence of viscosity on cellular behaviours is relatively unexplored and overlooked. Thus, in this study, the influence of viscosity on the adipogenic and osteogenic differentiation of human mesenchymal stem cells (hMSCs) was investigated by adjusting the viscosity of the culture medium. Viscosity exhibited different effects on adipogenic and osteogenic differentiation of hMSCs during two-dimensional (2D) culture. High viscosity facilitated osteogenic while inhibiting adipogenic differentiation. During adipogenic differentiation, the effect of viscosity on cell proliferation was negligible. However, during osteogenic differentiation, high viscosity decreased cell proliferation. The different influence of viscosity could be explained by the activation of mechanotransduction regulators of Yes-associated protein (YAP) and β-catenin. High viscosity could promote YAP and β-catenin nuclear translocation during osteogenic differentiation, which was responsible for the increased osteogenesis. High viscosity inhibited adipogenesis through promoting YAP nuclear translocation. This study could broaden the understanding of how viscosity can affect stem cell differentiation during 2D culture, which is valuable for tissue engineering.
Collapse
Affiliation(s)
- Chengyu Lu
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Tianjiao Zeng
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Man Wang
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Toru Yoshitomi
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
| | - Naoki Kawazoe
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
| | - Yingnan Yang
- Graduate School of Life and Environment Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| | - Guoping Chen
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
28
|
Ju H, Skibbe H, Fukui M, Yoshimura SH, Naoki H. Machine learning-guided reconstruction of cytoskeleton network from live-cell AFM images. iScience 2024; 27:110907. [PMID: 39391724 PMCID: PMC11465173 DOI: 10.1016/j.isci.2024.110907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Abstract
How actin filaments (F-actins) are dynamically reorganized in motile cells at the level of individual filaments is an open question. To find the answer, a high-speed atomic force microscopy (HS-AFM) system has been developed to live-imagine intracellular dynamics of the individual F-actins. However, noise and low resolution made it difficult to fully recognize individual F-actins in the HS-AFM images. To tackle this problem, we developed a new machine learning method that quantitatively recognizes individual F-actins. The method estimates F-actin orientation from the image while improving the resolution. We found that F-actins were oriented at ±35° toward the membrane in the lamellipodia, which is consistent with Arp2/3 complex-induced branching. Furthermore, in the cell cortex our results showed non-random orientation at four specific angles, suggesting a new mechanism for F-actin organization demonstrating the potential of our newly developed method to fundamentally improve our understanding of the structural dynamics of F-actin networks.
Collapse
Affiliation(s)
- Hanqiu Ju
- Laboratory of Data-driven Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan
- Mathematical Sciences Research Laboratory, Advanced Technology Research & Development Division, Nikon Corporation, 1-5-20, Nishioi, Shinagawa-ku, Tokyo 140-8601, Japan
| | - Henrik Skibbe
- Brain Image Analysis Unit, RIKEN Center for Brain Science, Wako-shi, Japan
| | - Masaya Fukui
- Laboratory of Data-driven Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Shige H. Yoshimura
- Laboratory of Plasma Membrane and Nuclear Signaling, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Center for Living Systems Information Science (CeLiSIS), Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Honda Naoki
- Laboratory of Data-driven Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan
- Graduate School of Medicine, Nagoya University, Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
- Theoretical Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| |
Collapse
|
29
|
Ohashi K, Kunitomi A, Chiba S, Mizuno K. Roles of the Dbl family of RhoGEFs in mechanotransduction - a review. Front Cell Dev Biol 2024; 12:1485725. [PMID: 39479515 PMCID: PMC11521908 DOI: 10.3389/fcell.2024.1485725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Rho guanine nucleotide exchange factors (RhoGEFs) comprise a wide range of proteins with a common domain responsible for the activation of the Rho family of small GTPases and various domains in other regions. The evolutionary divergence of RhoGEFs enables actin cytoskeletal reorganization, leading to complex cellular responses in higher organisms. In this review, we address the involvement of RhoGEFs in the mechanical stress response of mammalian cells. The cellular mechanical stress response is essential for the proper and orderly regulation of cell populations, including the maintenance of homeostasis, tissue morphogenesis, and adaptation to the mechanical environment. In particular, this review focuses on the recent findings regarding the Dbl family of RhoGEFs involved in mechanical stress responses at the cell-cell and cell-substrate adhesion sites, and their molecular mechanisms underlying actin cytoskeleton remodeling and signal transduction.
Collapse
Affiliation(s)
- Kazumasa Ohashi
- Department of Molecular and Chemical Life Sciences, Laboratory of Molecular and Cellular Biology, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | | | | | | |
Collapse
|
30
|
Horowitz A, Mammoto A, Sytnyk V, Jakovcevski I. Editorial: Editors' showcase 2023: insights in cell adhesion and migration. Front Cell Dev Biol 2024; 12:1497689. [PMID: 39421022 PMCID: PMC11483358 DOI: 10.3389/fcell.2024.1497689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Affiliation(s)
- Arie Horowitz
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Akiko Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Vladimir Sytnyk
- School of Biotech and Biomolecular Science, University of New South Wales, Kensington, NSW, Australia
| | - Igor Jakovcevski
- Department of Human Medicine, Faculty of Health, Witten-Herdecke University, Witten-Herdecke, Germany
| |
Collapse
|
31
|
Schwarz US. Cracking under stress: How actin might turn failure into action. Biophys J 2024; 123:3281-3282. [PMID: 39244639 PMCID: PMC11480752 DOI: 10.1016/j.bpj.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Affiliation(s)
- Ulrich S Schwarz
- Institute for Theoretical Physics and BioQuant, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
32
|
Shubhrasmita Sahu S, Sarkar P, Chattopadhyay A. Quantitation of F-actin in cytoskeletal reorganization: Context, methodology and implications. Methods 2024; 230:44-58. [PMID: 39074540 DOI: 10.1016/j.ymeth.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024] Open
Abstract
The actin cytoskeleton is involved in a large number of cellular signaling events in addition to providing structural integrity to the cell. Actin polymerization is a key event during cellular signaling. Although the role of actin cytoskeleton in cellular processes such as trafficking and motility has been extensively studied, the reorganization of the actin cytoskeleton upon signaling has been rarely explored due to lack of suitable assays. Keeping in mind this lacuna, we developed a confocal microscopy based approach that relies on high magnification imaging of cellular F-actin, followed by image reconstruction using commercially available software. In this review, we discuss the context and relevance of actin quantitation, followed by a detailed hands-on approach of the methodology involved with specific points on troubleshooting and useful precautions. In the latter part of the review, we elucidate the method by discussing applications of actin quantitation from our work in several important problems in contemporary membrane biology ranging from pathogen entry into host cells, to GPCR signaling and membrane-cytoskeleton interaction. We envision that future discovery of cell-permeable novel fluorescent probes, in combination with genetically encoded actin-binding reporters, would allow real-time visualization of actin cytoskeleton dynamics to gain deeper insights into active cellular processes in health and disease.
Collapse
Affiliation(s)
- Subhashree Shubhrasmita Sahu
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India; Department of Biochemistry, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Amitabha Chattopadhyay
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India.
| |
Collapse
|
33
|
Zhang P, Medwig-Kinney TN, Breiner EA, Perez JM, Song AN, Goldstein B. Cell signaling facilitates apical constriction by basolaterally recruiting Arp2/3 via Rac and WAVE. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614059. [PMID: 39386716 PMCID: PMC11463545 DOI: 10.1101/2024.09.23.614059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Apical constriction is a critical cell shape change that bends tissues. How precisely-localized actomyosin regulators drive apical constriction remains poorly understood. C. elegans gastrulation provides a valuable model to address this question. The Arp2/3 complex is essential in C. elegans gastrulation. To understand how Arp2/3 is locally regulated, we imaged embryos with endogenously-tagged Arp2/3 and its nucleation-promoting factors (NPFs). The three NPFs - WAVE, WASP, and WASH - colocalized with Arp2/3 and controlled Arp2/3 localization at distinct subcellular locations. We exploited this finding to study distinct populations of Arp2/3 and found that only WAVE depletion caused penetrant gastrulation defects. WAVE localized basolaterally with Arp2/3 at cell-cell contacts, dependent on CED-10/Rac. Establishing ectopic cell contacts recruited WAVE and Arp2/3, identifying contact as a symmetry-breaking cue for localization of these proteins. These results suggest that cell-cell signaling via Rac activates WAVE and Arp2/3 basolaterally, and that basolateral Arp2/3 is important for apical constriction.
Collapse
Affiliation(s)
- Pu Zhang
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Eleanor A. Breiner
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jadyn M. Perez
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - April N. Song
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bob Goldstein
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
34
|
Demosthene B, Kravchuk P, Harmon CL, Kalae A, Kang EH. Small organic osmolytes accelerate actin filament assembly and stiffen filaments. Cytoskeleton (Hoboken) 2024. [PMID: 39276026 DOI: 10.1002/cm.21927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 09/16/2024]
Abstract
Actin filament assembly and mechanics are crucial for maintenance of cell structure, motility, and division. Actin filament assembly occurs in a crowded intracellular environment consisting of various types of molecules, including small organic molecules known as osmolytes. Ample evidence highlights the protective functions of osmolytes such as trimethylamine-N-oxide (TMAO), including their effects on protein stability and their ability to counteract cellular osmotic stress. Yet, how TMAO affects individual actin filament assembly dynamics and mechanics is not well understood. We hypothesize that, owing to its protective nature, TMAO will enhance filament dynamics and stiffen actin filaments due to increased stability. In this study, we investigate osmolyte-dependent actin filament assembly and bending mechanics by measuring filament elongation rates, steady-state filament lengths, and bending persistence lengths in the presence of TMAO using total internal reflection fluorescence microscopy and pyrene assays. Our results demonstrate that TMAO increases filament elongation rates as well as steady-state average filament lengths, and enhances filament bending stiffness. Together, these results will help us understand how small organic osmolytes modulate cytoskeletal protein assembly and mechanics in living cells.
Collapse
Affiliation(s)
- Bryan Demosthene
- NanoScience Technology Center, University of Central Florida, Orlando, Florida, USA
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Pavlo Kravchuk
- NanoScience Technology Center, University of Central Florida, Orlando, Florida, USA
| | - Connor L Harmon
- NanoScience Technology Center, University of Central Florida, Orlando, Florida, USA
| | - Abdulrazak Kalae
- NanoScience Technology Center, University of Central Florida, Orlando, Florida, USA
| | - Ellen H Kang
- NanoScience Technology Center, University of Central Florida, Orlando, Florida, USA
- Department of Physics, University of Central Florida, Orlando, Florida, USA
- Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
35
|
Xu M, Rutkowski DM, Rebowski G, Boczkowska M, Pollard LW, Dominguez R, Vavylonis D, Ostap EM. Myosin-I synergizes with Arp2/3 complex to enhance the pushing forces of branched actin networks. SCIENCE ADVANCES 2024; 10:eado5788. [PMID: 39270022 PMCID: PMC11397503 DOI: 10.1126/sciadv.ado5788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024]
Abstract
Class I myosins (myosin-Is) colocalize with Arp2/3 complex-nucleated actin networks at sites of membrane protrusion and invagination, but the mechanisms by which myosin-I motor activity coordinates with branched actin assembly to generate force are unknown. We mimicked the interplay of these proteins using the "comet tail" bead motility assay, where branched actin networks are nucleated by the Arp2/3 complex on the surface of beads coated with myosin-I and nucleation-promoting factor. We observed that myosin-I increased bead movement efficiency by thinning actin networks without affecting growth rates. Myosin-I triggered symmetry breaking and comet tail formation in dense networks resistant to spontaneous fracturing. Even with arrested actin assembly, myosin-I alone could break the network. Computational modeling recapitulated these observations, suggesting myosin-I acts as a repulsive force shaping the network's architecture and boosting its force-generating capacity. We propose that myosin-I leverages its power stroke to amplify the forces generated by Arp2/3 complex-nucleated actin networks.
Collapse
Affiliation(s)
- Mengqi Xu
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Grzegorz Rebowski
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Malgorzata Boczkowska
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Luther W. Pollard
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roberto Dominguez
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - E. Michael Ostap
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
36
|
Boiero Sanders M, Oosterheert W, Hofnagel O, Bieling P, Raunser S. Phalloidin and DNase I-bound F-actin pointed end structures reveal principles of filament stabilization and disassembly. Nat Commun 2024; 15:7969. [PMID: 39261469 PMCID: PMC11390976 DOI: 10.1038/s41467-024-52251-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Actin filament turnover involves subunits binding to and dissociating from the filament ends, with the pointed end being the primary site of filament disassembly. Several molecules modulate filament turnover, but the underlying mechanisms remain incompletely understood. Here, we present three cryo-EM structures of the F-actin pointed end in the presence and absence of phalloidin or DNase I. The two terminal subunits at the undecorated pointed end adopt a twisted conformation. Phalloidin can still bind and bridge these subunits, inducing a conformational shift to a flattened, F-actin-like state. This explains how phalloidin prevents depolymerization at the pointed end. Interestingly, two DNase I molecules simultaneously bind to the phalloidin-stabilized pointed end. In the absence of phalloidin, DNase I binding would disrupt the terminal actin subunit packing, resulting in filament disassembly. Our findings uncover molecular principles of pointed end regulation and provide structural insights into the kinetic asymmetry between the actin filament ends.
Collapse
Affiliation(s)
- Micaela Boiero Sanders
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Wout Oosterheert
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Oliver Hofnagel
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany.
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany.
| |
Collapse
|
37
|
Paul S, Gayen K, Cantavella PG, Escuder B, Singh N. Complex Pathways Drive Pluripotent Fmoc-Leucine Self-Assemblies. Angew Chem Int Ed Engl 2024; 63:e202406220. [PMID: 38825832 DOI: 10.1002/anie.202406220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/25/2024] [Accepted: 05/31/2024] [Indexed: 06/04/2024]
Abstract
Nature uses complex self-assembly pathways to access distinct functional non-equilibrium self-assemblies. This remarkable ability to steer same set of biomolecules into different self-assembly states is done by avoiding thermodynamic pit. In synthetic systems, on demand control over 'Pathway Complexity' to access self-assemblies different from equilibrium structures remains challenging. Here we show versatile non-equilibrium assemblies of the same monomer via alternate assembly pathways. The assemblies nucleate using non-classical or classical nucleation routes into distinct metastable (transient hydrogels), kinetic (stable hydrogels) and thermodynamic structures [(poly)-crystals and 2D sheets]. Initial chemical and thermal inputs force the monomers to follow different assembly pathways and form soft-materials with distinct molecular arrangements than at equilibrium. In many cases, equilibrium structures act as thermodynamic sink which consume monomers from metastable structures giving transiently formed materials. This dynamics can be tuned chemically or thermally to slow down the dissolution of transient hydrogel, or skip the intermediate hydrogel altogether to reach final equilibrium assemblies. If required this metastable state can be kinetically trapped to give strong hydrogel stable over days. This method to control different self-assembly states can find potential use in similar biomimetic systems to access new materials for various applications.
Collapse
Affiliation(s)
- Subir Paul
- Institute of Advanced Materials, Universitat Jaume I, Avinguda de Vicent Sos Baynat, s/n, 12006, Castelló de la Plana, Castelló, Spain
| | - Kousik Gayen
- Institute of Advanced Materials, Universitat Jaume I, Avinguda de Vicent Sos Baynat, s/n, 12006, Castelló de la Plana, Castelló, Spain
| | - Pau Gil Cantavella
- Institute of Advanced Materials, Universitat Jaume I, Avinguda de Vicent Sos Baynat, s/n, 12006, Castelló de la Plana, Castelló, Spain
| | - Beatriu Escuder
- Institute of Advanced Materials, Universitat Jaume I, Avinguda de Vicent Sos Baynat, s/n, 12006, Castelló de la Plana, Castelló, Spain
| | - Nishant Singh
- Institute of Advanced Materials, Universitat Jaume I, Avinguda de Vicent Sos Baynat, s/n, 12006, Castelló de la Plana, Castelló, Spain
| |
Collapse
|
38
|
Limatola N, Chun JT, Schmitt JL, Lehn JM, Santella L. The Effect of Synthetic Polyamine BPA-C8 on the Fertilization Process of Intact and Denuded Sea Urchin Eggs. Cells 2024; 13:1477. [PMID: 39273047 PMCID: PMC11394060 DOI: 10.3390/cells13171477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Sea urchin eggs are covered with layers of extracellular matrix, namely, the vitelline layer (VL) and jelly coat (JC). It has been shown that sea urchin eggs' JC components serve as chemoattractants or ligands for the receptor on the fertilizing sperm to promote the acrosome reaction. Moreover, the egg's VL provides receptors for conspecific sperm to bind, and, to date, at least two sperm receptors have been identified on the surface of sea urchin eggs. Interestingly, however, according to our previous work, denuded sea urchin eggs devoid of the JC and VL do not fail to become fertilized by sperm. Instead, they are bound and penetratedby multiple sperm, raising the possibility that an alternative pathway independent of the VL-residing sperm receptor may be at work. In this research, we studied the roles of the JC and VL using intact and denuded eggs and the synthetic polyamine BPA-C8. BPA-C8 is known to bind to the negatively charged macromolecular complexes in the cells, such as the JC, VL, and the plasma membrane of echinoderm eggs, as well as to the actin filaments in fibroblasts. Our results showed that, when added to seawater, BPA-C8 significantly repressed the Ca2+ wave in the intact P. lividus eggs at fertilization. In eggs deprived of the VL and JC, BPA-C8 binds to the plasma membrane and increases fibrous structures connecting microvilli, thereby allowing the denuded eggs to revert towards monospermy at fertilization. However, the reduced Ca2+ signal in denuded eggs was nullified compared to the intact eggs because removing the JC and VL already decreased the Ca2+ wave. BPA-C8 does not cross the VL and the cell membrane of unfertilized sea urchin eggs to diffuse into the cytoplasm at variance with the fibroblasts. Indeed, the jasplakinolide-induced polymerization of subplasmalemmal actin filaments was inhibited in the eggs microinjected with BPA-C8, but not in the ones bath-incubated with the same dose of BPA-C8.
Collapse
Affiliation(s)
- Nunzia Limatola
- Department of Research Infrastructures for Marine Biological Resources, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| | - Jong Tai Chun
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| | - Jean-Louis Schmitt
- Laboratory of Supramolecular Chemistry, Institut de Science et d'Ingénierie Supramoléculaires ISIS, Université de Strasbourg, 8 Allée Gaspard Monge, 67000 Strasbourg, France
| | - Jean-Marie Lehn
- Laboratory of Supramolecular Chemistry, Institut de Science et d'Ingénierie Supramoléculaires ISIS, Université de Strasbourg, 8 Allée Gaspard Monge, 67000 Strasbourg, France
| | - Luigia Santella
- Department of Research Infrastructures for Marine Biological Resources, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| |
Collapse
|
39
|
Gupta R, Dittmeier M, Wohlleben G, Nickl V, Bischler T, Luzak V, Wegat V, Doll D, Sodmann A, Bady E, Langlhofer G, Wachter B, Havlicek S, Gupta J, Horn E, Lüningschrör P, Villmann C, Polat B, Wischhusen J, Monoranu CM, Kuper J, Blum R. Atypical cellular responses mediated by intracellular constitutive active TrkB (NTRK2) kinase domains and a solely intracellular NTRK2-fusion oncogene. Cancer Gene Ther 2024; 31:1357-1379. [PMID: 39039193 PMCID: PMC11405271 DOI: 10.1038/s41417-024-00809-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 06/25/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024]
Abstract
Trk (NTRK) receptor and NTRK gene fusions are oncogenic drivers of a wide variety of tumors. Although Trk receptors are typically activated at the cell surface, signaling of constitutive active Trk and diverse intracellular NTRK fusion oncogenes is barely investigated. Here, we show that a high intracellular abundance is sufficient for neurotrophin-independent, constitutive activation of TrkB kinase domains. In HEK293 cells, constitutive active TrkB kinase and an intracellular NTRK2-fusion oncogene (SQSTM1-NTRK2) reduced actin filopodia dynamics, phosphorylated FAK, and altered the cell morphology. Atypical cellular responses could be mimicked with the intracellular kinase domain, which did not activate the Trk-associated MAPK/ERK pathway. In glioblastoma-like U87MG cells, expression of TrkB or SQSTM1-NTRK2 reduced cell motility and caused drastic changes in the transcriptome. Clinically approved Trk inhibitors or mutating Y705 in the kinase domain, blocked the cellular effects and transcriptome changes. Atypical signaling was also seen for TrkA and TrkC. Moreover, hallmarks of atypical pTrk kinase were found in biopsies of Nestin-positive glioblastoma. Therefore, we suggest Western blot-like immunoassay screening of NTRK-related (brain) tumor biopsies to identify patients with atypical panTrk or phosphoTrk signals. Such patients could be candidates for treatment with NTRK inhibitors such as Larotrectinhib or Entrectinhib.
Collapse
Affiliation(s)
- Rohini Gupta
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Melanie Dittmeier
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Gisela Wohlleben
- Department of Radiation Oncology, University of Würzburg, Würzburg, Germany
| | - Vera Nickl
- Department of Neurosurgery, Section Experimental Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Thorsten Bischler
- Core Unit Systems Medicine, University of Würzburg, Würzburg, Germany
| | - Vanessa Luzak
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
- Ludwig-Maximilians-Universität München, Biomedizinisches Zentrum, Planegg, Germany
| | - Vanessa Wegat
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
- Fraunhofer-Institut für Grenzflächen- und Bioverfahrenstechnik IGB, Bio- Elektro- und Chemokatalyse BioCat, Straubing, Germany
| | - Dennis Doll
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Annemarie Sodmann
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Elena Bady
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Georg Langlhofer
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Britta Wachter
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Steven Havlicek
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
- Neurona Therapeutics, 170 Harbor Way, South San Francisco, CA, USA
| | - Jahnve Gupta
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Evi Horn
- Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Patrick Lüningschrör
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Carmen Villmann
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Bülent Polat
- Department of Radiation Oncology, University of Würzburg, Würzburg, Germany
| | - Jörg Wischhusen
- Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Camelia M Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Jochen Kuper
- Rudolf Virchow Center for Experimental Biomedicine, Institute for Structural Biology, University of Würzburg, Würzburg, Germany
| | - Robert Blum
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany.
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
40
|
deWeever A, Paudel SS, Zhou C, Francis CM, Tambe DT, Frank DW, Balczon R, Stevens T. cUMP elicits interendothelial gap formation during Pseudomonas aeruginosa infection. Am J Physiol Lung Cell Mol Physiol 2024; 327:L395-L405. [PMID: 39076085 PMCID: PMC11444506 DOI: 10.1152/ajplung.00164.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 05/08/2024] [Accepted: 06/30/2024] [Indexed: 07/31/2024] Open
Abstract
Pseudomonas aeruginosa utilizes a type 3 secretion system to intoxicate host cells with the nucleotidyl cyclase ExoY. After activation by its host cell cofactor, filamentous actin, ExoY produces purine and pyrimidine cyclic nucleotides, including cAMP, cGMP, and cUMP. ExoY-generated cyclic nucleotides promote interendothelial gap formation, impair motility, and arrest cell growth. The disruptive activities of cAMP and cGMP during the P. aeruginosa infection are established; however, little is known about the function of cUMP. Here, we tested the hypothesis that cUMP contributes to endothelial cell barrier disruption during P. aeruginosa infection. Using a membrane permeable cUMP analog, cUMP-AM, we revealed that during infection with catalytically inactive ExoY, cUMP promotes interendothelial gap formation in cultured pulmonary microvascular endothelial cells (PMVECs) and contributes to increased filtration coefficient in the isolated perfused lung. These findings indicate that cUMP contributes to endothelial permeability during P. aeruginosa lung infection.NEW & NOTEWORTHY During pneumonia, bacteria utilize a virulence arsenal to communicate with host cells. The Pseudomonas aeruginosa T3SS directly introduces virulence molecules into the host cell cytoplasm. These molecules are enzymes that trigger interkingdom communication. One of the exoenzymes is a nucleotidyl cyclase that produces noncanonical cyclic nucleotides like cUMP. Little is known about how cUMP acts in the cell. Here we found that cUMP instigates pulmonary edema during Pseudomonas aeruginosa infection of the lung.
Collapse
Grants
- R01 HL167997 NHLBI NIH HHS
- HL136689 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL066299 NHLBI NIH HHS
- AI104922 HHS | NIH | NIAID | Division of Microbiology and Infectious Diseases (DMID)
- R01 HL140182 NHLBI NIH HHS
- HL167997 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 AI104922 NIAID NIH HHS
- HL148069 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL148069 NHLBI NIH HHS
- HL140182 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL66299 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Althea deWeever
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Sunita S Paudel
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Chun Zhou
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - C Michael Francis
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Dhananjay T Tambe
- Department of Mechanical, Aerospace and Biomedical Engineering, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Dara W Frank
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
41
|
Xiong H, Cao M, Yu Y, Duan X, Sun L, Tang L, Fan X. Study on the Effects of Low-Intensity Pulsed Ultrasound and Iron Ions for Proliferation and Differentiation of Osteoblasts. ULTRASOUND IN MEDICINE & BIOLOGY 2024:S0301-5629(24)00265-5. [PMID: 39209558 DOI: 10.1016/j.ultrasmedbio.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE This study involved the proliferation and differentiation of osteoblasts treated with low-intensity pulsed ultrasound (LIPUS) and iron (Fe3+) ions, respectively. The biological effects of LIPUS and Fe3+ ions on the proliferation and differentiation of osteoblasts were also evaluated. METHODS MC3T3-E1 cells were seeded in six-well plates with the medium, which contained different concentrations of Fe3+ (0, 100, 200, 300, 400, 500, 600 and 700 μg L-1, respectively). LIPUS treatment was directed at the bottom of the plate for 20 min at an intensity of 80 mW cm-2 every day. RESULTS Viability results showed that a dose of 400 μg L-1 Fe3+ ions had the best effect at promoting osteogenic proliferation in cell culture. The results of alkaline phosphatase staining and mineralization indicated that the differentiation of osteoblasts was promoted by LIPUS and Fe3+ ions. Fluorescence staining results showed that the number of cell nuclei in the LIPUS, Fe3+ and LIPUS-Fe groups increased by 37.20%, 55.81% and 89.76%, respectively. Migration data indicated that migration and proliferation rates were increased by LIPUS and Fe3+, and the results of protein expression indicated that LIPUS and Fe3+ may increase the expression of Wnt, β-catenin, and Runx2, hence promoting normal bone regeneration and development. CONCLUSION The combination of LIPUS (1.5 MHz, 80 mW cm-2) and Fe3+ accelerates the proliferation and differentiation of osteoblasts significantly compared with single-factor treatment (stimulated by LIPUS and Fe3+ ions, respectively). This study could establish a foundation for LIPUS-responsive biomaterials in the repair and regeneration of bone tissues.
Collapse
Affiliation(s)
- Huanbin Xiong
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Mengshu Cao
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Yanan Yu
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Xueyou Duan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China.
| |
Collapse
|
42
|
Jaeger HM, Murugan A, Nagel SR. Training physical matter to matter. SOFT MATTER 2024; 20:6695-6701. [PMID: 39140794 DOI: 10.1039/d4sm00629a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Biological systems offer a great many examples of how sophisticated, highly adapted behavior can emerge from training. Here we discuss how training might be used to impart similarly adaptive properties in physical matter. As a special form of materials processing, training differs in important ways from standard approaches of obtaining sought after material properties. In particular, rather than designing or programming the local configurations and interactions of constituents, training uses externally applied stimuli to evolve material properties. This makes it possible to obtain different functionalities from the same starting material (pluripotency). Furthermore, training evolves a material in situ or under conditions similar to those during the intended use; thus, material performance can improve rather than degrade over time. We discuss requirements for trainability, outline recently developed training strategies for creating soft materials with multiple, targeted and adaptable functionalities, and provide examples where the concept of training has been applied to materials on length scales from the molecular to the macroscopic.
Collapse
Affiliation(s)
- Heinrich M Jaeger
- The James Franck Institute and Department of Physics, The University of Chicago, 929 E 57th St., Chicago, Illinois 60637, USA.
| | - Arvind Murugan
- The James Franck Institute and Department of Physics, The University of Chicago, 929 E 57th St., Chicago, Illinois 60637, USA.
| | - Sidney R Nagel
- The James Franck Institute and Department of Physics, The University of Chicago, 929 E 57th St., Chicago, Illinois 60637, USA.
| |
Collapse
|
43
|
Dong J, Zhu XN, Zeng PM, Cao DD, Yang Y, Hu J, Luo ZG. A hominoid-specific signaling axis regulating the tempo of synaptic maturation. Cell Rep 2024; 43:114548. [PMID: 39052482 DOI: 10.1016/j.celrep.2024.114548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/15/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Human cortical neurons (hCNs) exhibit high dendritic complexity and synaptic density, and the maturation process is greatly protracted. However, the molecular mechanism governing these specific features remains unclear. Here, we report that the hominoid-specific gene TBC1D3 promotes dendritic arborization and protracts the pace of synaptogenesis. Ablation of TBC1D3 in induced hCNs causes reduction of dendritic growth and precocious synaptic maturation. Forced expression of TBC1D3 in the mouse cortex protracts synaptic maturation while increasing dendritic growth. Mechanistically, TBC1D3 functions via interaction with MICAL1, a monooxygenase that mediates oxidation of actin filament. At the early stage of differentiation, the TBC1D3/MICAL1 interaction in the cytosol promotes dendritic growth via F-actin oxidation and enhanced actin dynamics. At late stages, TBC1D3 escorts MICAL1 into the nucleus and downregulates the expression of genes related with synaptic maturation through interaction with the chromatin remodeling factor ATRX. Thus, this study delineates the molecular mechanisms underlying human neuron development.
Collapse
Affiliation(s)
- Jian Dong
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Xiao-Na Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Peng-Ming Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Dong-Dong Cao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yang Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhen-Ge Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
44
|
Valera JS, López-Acosta Á, Hermans TM. Photoinitiated Transient Self-Assembly in a Catalytically Driven Chemical Reaction Cycle. Angew Chem Int Ed Engl 2024; 63:e202406931. [PMID: 38770670 DOI: 10.1002/anie.202406931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 05/22/2024]
Abstract
Chemically fueled chemical reaction networks (CRNs) are essential in controlling dissipative self-assembly. A key challenge in the field is to store chemical fuel-precursors or "pre-fuels" in the system that are converted into activating or deactivating fuels in a catalytically controlled CRN. In addition, real-time control over catalysis in a CRN by light is highly desirable, but so far not yet achieved. Here we show a catalytically driven CRN that is photoinitiated with 450 nm light, producing activated monomers that go on to perform transient self-assembly. Monomer activation proceeds via photoredox catalysis, converting the monomer alcohol groups into the corresponding aldehydes that self-assemble into large supramolecular fibers. Monomer deactivation is achieved by organometallic catalysis that relies on pre-fuel hydrolysis to release formate (i.e. the deactivating fuel). Additionally, irradiation with 305 nm light accelerates the release of formate by photo-uncaging the pre-fuel, leading to a factor of ca. 2 faster deactivation of the monomer. Overall, we show transient self-assembly upon visible light photoactivation, and tunable life-times by ultraviolet light.
Collapse
Affiliation(s)
- Jorge S Valera
- IMDEA Nanociencia, C/ Faraday 9, 28049, Madrid, Spain
- Université de Strasbourg, CNRS, UMR7140, 4 Rue Blaise Pascal, 67081, Strasbourg, France
| | | | - Thomas M Hermans
- IMDEA Nanociencia, C/ Faraday 9, 28049, Madrid, Spain
- Université de Strasbourg, CNRS, UMR7140, 4 Rue Blaise Pascal, 67081, Strasbourg, France
| |
Collapse
|
45
|
Salvadori A, Bonanno C, Serpelloni M, McMeeking RM. On the generation of force required for actin-based motility. Sci Rep 2024; 14:18384. [PMID: 39117762 PMCID: PMC11310465 DOI: 10.1038/s41598-024-69422-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
The fundamental question of how forces are generated in a motile cell, a lamellipodium, and a comet tail is the subject of this note. It is now well established that cellular motility results from the polymerization of actin, the most abundant protein in eukaryotic cells, into an interconnected set of filaments. We portray this process in a continuum mechanics framework, claiming that polymerization promotes a mechanical swelling in a narrow zone around the nucleation loci, which ultimately results in cellular or bacterial motility. To this aim, a new paradigm in continuum multi-physics has been designed, departing from the well-known theory of Larché-Cahn chemo-transport-mechanics. In this note, we set up the theory of network growth and compare the outcomes of numerical simulations with experimental evidence.
Collapse
Affiliation(s)
- Alberto Salvadori
- The Mechanobiology Research Center, UNIBS, 25123, Brescia, Italy.
- Department of Mechanical and Industrial Engineering, Università degli Studi di Brescia, via Branze 38, 25123, Brescia, Italy.
| | - Claudia Bonanno
- The Mechanobiology Research Center, UNIBS, 25123, Brescia, Italy
| | - Mattia Serpelloni
- The Mechanobiology Research Center, UNIBS, 25123, Brescia, Italy
- Department of Mechanical and Industrial Engineering, Università degli Studi di Brescia, via Branze 38, 25123, Brescia, Italy
| | - Robert M McMeeking
- The Mechanobiology Research Center, UNIBS, 25123, Brescia, Italy
- Materials and Mechanical Engineering Departments, University of California, Santa Barbara, CA, 93106, USA
- School of Engineering, University of Aberdeen, King's College, Aberdeen, AB24 3UE, UK
- INM - Leibniz Institute for New Materials, Campus D2 2, Saarbruecken, Germany
| |
Collapse
|
46
|
Wubshet NH, Young CJ, Liu AP. Rearrangement of GUV-confined actin networks in response to micropipette aspiration. Cytoskeleton (Hoboken) 2024; 81:310-317. [PMID: 38326972 PMCID: PMC11303591 DOI: 10.1002/cm.21836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
Although diverse actin network architectures found inside the cell have been individually reconstituted outside of the cell, how different types of actin architectures reorganize under applied forces is not entirely understood. Recently, bottom-up reconstitution has enabled studies where dynamic and phenotypic characteristics of various actin networks can be recreated in an isolated cell-like environment. Here, by creating a giant unilamellar vesicle (GUV)-based cell model encapsulating actin networks, we investigate how actin networks rearrange in response to localized stresses applied by micropipette aspiration. We reconstitute actin bundles and branched bundles in GUVs separately and mechanically perturb them. Interestingly, we find that, when aspirated, protrusive actin bundles that are otherwise randomly oriented in the GUV lumen collapse and align along the axis of the micropipette. However, when branched bundles are aspirated, the network remains intact and outside of the pipette while the GUV membrane is aspirated into the micropipette. These results reveal distinct responses in the rearrangement of actin networks in a network architecture-dependent manner when subjected to physical forces.
Collapse
Affiliation(s)
- Nadab H. Wubshet
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Cole J. Young
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Allen P. Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
47
|
Mukadum F, Ccoa WJP, Hocky GM. Molecular simulation approaches to probing the effects of mechanical forces in the actin cytoskeleton. Cytoskeleton (Hoboken) 2024; 81:318-327. [PMID: 38334204 PMCID: PMC11310368 DOI: 10.1002/cm.21837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024]
Abstract
In this article we give our perspective on the successes and promise of various molecular and coarse-grained simulation approaches to probing the effect of mechanical forces in the actin cytoskeleton.
Collapse
Affiliation(s)
- Fatemah Mukadum
- Department of Chemistry, New York University, New York, NY 10003, USA
| | | | - Glen M. Hocky
- Department of Chemistry, New York University, New York, NY 10003, USA
- Simons Center for Computational Physical Chemistry, New York, NY 10003, USA
| |
Collapse
|
48
|
Read TA, Cisterna BA, Skruber K, Ahmadieh S, Liu TM, Vitriol JA, Shi Y, Black JB, Butler MT, Lindamood HL, Lefebvre AE, Cherezova A, Ilatovskaya DV, Bear JE, Weintraub NL, Vitriol EA. The actin binding protein profilin 1 localizes inside mitochondria and is critical for their function. EMBO Rep 2024; 25:3240-3262. [PMID: 39026010 PMCID: PMC11316047 DOI: 10.1038/s44319-024-00209-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 06/16/2024] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
The monomer-binding protein profilin 1 (PFN1) plays a crucial role in actin polymerization. However, mutations in PFN1 are also linked to hereditary amyotrophic lateral sclerosis, resulting in a broad range of cellular pathologies which cannot be explained by its primary function as a cytosolic actin assembly factor. This implies that there are important, undiscovered roles for PFN1 in cellular physiology. Here we screened knockout cells for novel phenotypes associated with PFN1 loss of function and discovered that mitophagy was significantly upregulated. Indeed, despite successful autophagosome formation, fusion with the lysosome, and activation of additional mitochondrial quality control pathways, PFN1 knockout cells accumulate depolarized, dysmorphic mitochondria with altered metabolic properties. Surprisingly, we also discovered that PFN1 is present inside mitochondria and provide evidence that mitochondrial defects associated with PFN1 loss are not caused by reduced actin polymerization in the cytosol. These findings suggest a previously unrecognized role for PFN1 in maintaining mitochondrial integrity and highlight new pathogenic mechanisms that can result from PFN1 dysregulation.
Collapse
Affiliation(s)
- Tracy-Ann Read
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Bruno A Cisterna
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Kristen Skruber
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Samah Ahmadieh
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Tatiana M Liu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Josefine A Vitriol
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Yang Shi
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Population Health Sciences, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Joseph B Black
- Division of Urologic Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Mitchell T Butler
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Halli L Lindamood
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | | | - Alena Cherezova
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Eric A Vitriol
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
49
|
Banerjee DS, Freedman SL, Murrell MP, Banerjee S. Growth-induced collective bending and kinetic trapping of cytoskeletal filaments. Cytoskeleton (Hoboken) 2024; 81:409-419. [PMID: 38775207 DOI: 10.1002/cm.21877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024]
Abstract
Growth and turnover of actin filaments play a crucial role in the construction and maintenance of actin networks within cells. Actin filament growth occurs within limited space and finite subunit resources in the actin cortex. To understand how filament growth shapes the emergent architecture of actin networks, we developed a minimal agent-based model coupling filament mechanics and growth in a limiting subunit pool. We find that rapid filament growth induces kinetic trapping of highly bent actin filaments. Such collective bending patterns are long-lived, organized around nematic defects, and arise from competition between filament polymerization and bending elasticity. The stability of nematic defects and the extent of kinetic trapping are amplified by an increase in the abundance of the actin pool and by crosslinking the network. These findings suggest that kinetic trapping is a robust consequence of growth in crowded environments, providing a route to program shape memory in actin networks.
Collapse
Affiliation(s)
- Deb Sankar Banerjee
- Department of Physics, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
- James Franck Institute, University of Chicago, Chicago, Illinois, USA
| | | | - Michael P Murrell
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Systems Biology Institute, West Haven, Connecticut, USA
- Department of Physics, Yale University, New Haven, Connecticut, USA
| | - Shiladitya Banerjee
- Department of Physics, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
50
|
Xu XP, Cao W, Swift MF, Pandit NG, Huehn AE, Sindelar CV, De La Cruz EM, Hanein D, Volkmann N. High-resolution yeast actin structures indicate the molecular mechanism of actin filament stiffening by cations. Commun Chem 2024; 7:164. [PMID: 39079963 PMCID: PMC11289367 DOI: 10.1038/s42004-024-01243-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Actin filament assembly and the regulation of its mechanical properties are fundamental processes essential for eukaryotic cell function. Residue E167 in vertebrate actins forms an inter-subunit salt bridge with residue K61 of the adjacent subunit. Saccharomyces cerevisiae actin filaments are more flexible than vertebrate filaments and have an alanine at this position (A167). Substitution of this alanine for a glutamic acid (A167E) confers Saccharomyces cerevisiae actin filaments with salt-dependent stiffness similar to vertebrate actins. We developed an optimized cryogenic electron microscopy workflow refining sample preparation and vitrification to obtain near-atomic resolution structures of wild-type and A167E mutant Saccharomyces cerevisiae actin filaments. The difference between these structures allowed us to pinpoint the potential binding site of a filament-associated cation that controls the stiffness of the filaments in vertebrate and A167E Saccharomyces cerevisiae actins. Through an analysis of previously published high-resolution reconstructions of vertebrate actin filaments, along with a newly determined high-resolution vertebrate actin structure in the absence of potassium, we identified a unique peak near residue 167 consistent with the binding of a magnesium ion. Our findings show how magnesium can contribute to filament stiffening by directly bridging actin subunits and allosterically affecting the orientation of the DNase-I binding loop of actin, which plays a regulatory role in modulating actin filament stiffness and interactions with regulatory proteins.
Collapse
Affiliation(s)
- Xiao-Ping Xu
- Scintillon Institute, 6868 Nancy Ridge Drive, San Diego, CA, 92121, USA
| | - Wenxiang Cao
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA
| | - Mark F Swift
- Scintillon Institute, 6868 Nancy Ridge Drive, San Diego, CA, 92121, USA
| | - Nandan G Pandit
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA
| | - Andrew E Huehn
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA
| | - Charles V Sindelar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA
| | - Enrique M De La Cruz
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA
| | - Dorit Hanein
- Department of Chemistry and Biochemistry and Department of Biological Engineering, University of California, Santa Barbara, CA, 93106, USA.
| | - Niels Volkmann
- Department of Biological Engineering, Department of Electrical and Computer Engineering, Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA, 93106, USA.
| |
Collapse
|